Skip to main content

ORIGINAL RESEARCH article

Front. Oncol., 09 September 2020
Sec. Hematologic Malignancies

Aberrant Chloride Intracellular Channel 4 Expression Is Associated With Adverse Outcome in Cytogenetically Normal Acute Myeloid Leukemia

  • 1Department of Hematology, First Medical Center, Chinese PLA General Hospital, Beijing, China
  • 2School of Electrical and Computer Engineering, Purdue University, West Lafayette, IN, United States
  • 3Department of Emergency, First Medical Center, Chinese PLA General Hospital, Beijing, China

Background and Methods: Acute myeloid leukemia (AML), which starts in the bone marrow, is a group of hematopoietic stem cell disorders. Chloride intracellular channel 4 (CLIC4) is regulated by p53, c-Myc, and TGF-β. It induces the NF-κB-dependent activation of HIF (hypoxia-inducible factor) and participates in tumor growth through its microenvironmental function. However, its prognostic value in AML remains unclear, as well as its co-expression biomarkers. In this study, we evaluated the prognostic significance of CLIC4 expression using two independent large cohorts of cytogenetically normal AML (CN-AML) patients. Multivariable analysis and multi-omics analysis with weighted correlation network analysis (WGCNA) in the CN-AML group were also presented. Based on CLIC4 and its related genes, microRNA–target gene interaction network analysis and downstream gene ontology analysis were performed to unveil the complex functions behind CLIC4.

Results: We demonstrated that the overexpression of CLIC4 was notably associated with unfavorable outcome in the two independent cohorts of CN-AML patients [overall survival (OS) and event-free survival (EFS): P < 0.0001, n = 185; OS: P = 0.016, n = 232], as well as in the European LeukemiaNet (ELN) Intermediate-I group (OS: P = 0.015, EFS: P = 0.012, n = 115), the National Comprehensive Cancer Network Intermediate Risk AML group (OS and EFS: P < 0.0001, n = 225), and the non-M3 AML group (OS and EFS: P < 0.0001, n = 435). Multivariable analysis further validated CLIC4 as a high-risk factor in the CN-AML group. Multi-omics analysis presented the overexpression of CLIC4 as associated with the co-expression of the different gene sets in leukemia, up/downregulation of the immune-related pathways, dysregulation of microRNAs, and hypermethylation around the CpG islands, in open sea regions, and in different gene structural fragments including TSS1500, gene body, 5′UTR region, 3′UTR region, and the first exon. By further performing WGCNA on multi-omics data, certain biomarkers that are co-expressed with CLIC4 were also unveiled.

Conclusion: We demonstrated that CLIC4 is a novel, potential unfavorable prognosticator and therapeutic target for CN-AML. As having a key role in CN-AML, the interactions between CLIC4 and other genomics and transcriptomics data were confirmed by performing microRNA–target gene interaction network analysis and gene ontology enrichment analysis. The experimental result provides evidence for the clinical strategy selection of CN-AML patients.

Introduction

Acute myeloid leukemia (AML) is a genetically and clinically heterogeneous hematopoietic malignancy. The outcome of AML patients is generally poor and is strongly influenced by gene mutations and chromosomal alterations (1). Currently, the prognostic stratification is based on gene sequencing and cytogenetic analysis. It is used clinically to guide treatment decisions in AML patients with chromosomal abnormalities, which comprises 55–60% of all AML cases (2). However, the risk stratification guidelines for the remaining 40–45% AML patients, who lack detectable chromosomal alternations and are classified as cytogenetically normal AML (CN-AML) patients, are still not well understood (3). For CN-AML patients, the outcomes are widely diverse (4). According to the 2017 European LeukemiaNet (ELN) AML risk stratification, it is entirely determined by common gene mutations such as NPM1, ASXL1, TP53, RUNX1, FLT3-ITD, double mutation of CEBPA, etc. (5). Although the clinical validation of these molecular markers has added a great deal to the prognostic stratification of CN-AML, continuing to elucidate the biological characteristics is vital to guide treatment and provide novel targets for therapies. Accurately predicting the therapy outcome and establishing a precise risk stratification have therefore become crucial for CN-AML.

Over the past decade, the biological and physiological role for ion channels in tumor development has been widely studied. Ion channels are integral membrane proteins preserving the normal physiology of cells and regulating many cellular, organellar, and physiological processes. Evidence reveals the multitask possibility of the participation of ion channel proteins in the tumorigenesis and leukemogenesis process (6). The abnormal expression and function of ion channel proteins can contribute to the development, metastasis, and drug resistance of leukemia (7). Many investigations have indicated that the overexpression of some ion channel proteins is related to poor prognosis. Therefore, ion channels could be considered as new biomarkers in leukemia diagnosis and treatment (8).

Chloride intracellular ion channels (CLICs), a novel class of intracellular organelle ion channels, are now being widely surveyed to explore their role in tumor biology. When overexpressed heterologously, the CLIC protein family is able to generate an outwardly rectifying chloride channel activity (9). Among the CLIC protein family (CLIC1–CLIC6), several studies have focused on the role of chloride intracellular channel 4 (CLIC4) protein. In soluble form, CLIC4 is involved in p53- and c-myc-mediated apoptosis and transforming growth factor beta (TGF-β) signaling modulation in multiple cell types (10). CLIC4 has been proven to be involved in multiple cellular functions, including the regulation of cell proliferation, differentiation, angiogenesis, and apoptosis (11). Studies implied that CLIC4 also induced the NF-κB-dependent activation of HIF (hypoxia-inducible factor) and participated in tumor growth through its microenvironmental function (11, 12). Subsequent studies found that CLIC4 might be associated with adverse outcomes in colorectal cancer and esophageal squamous cell carcinoma (13, 14). Specifically, an elevated expression of CLIC4, a common trait of many cancers and a marker of malignant progression, was found to be associated with the activation of the Arf6 and NF-κB signaling pathway (15). However, its prognostic value in AML remains unclear.

Here, we presented CLIC4 as a novel, potential unfavorable prognosticator and therapeutic target for CN-AML. We also explored CLIC4-associated genomic and epigenomic patterns to further elucidate its key role in CN-AML. Our study represented direct evidences for CLIC4 expression as a prognostic biomarker in multiple CN-AML datasets. We also brought insights into CN-AML risk stratification and potential therapeutic targets in leukemia prognosis and treatment.

Materials and Methods

Patients and Treatment

This study was approved by local institutional review boards and written informed consent was provided for all patients according to the Declaration of Helsinki.

The first cohort was derived from the whole AML cohort (GSE6891) with 185 primarily untreated CN-AML patients (median age = 47 years, range = 16–60 years) diagnosed and collected at Erasmus University Medical Center from 1990 to 2008. Conventional cytogenetics examined more than 20 metaphases from bone marrow (BM) samples to determine normal karyotype.

The second independent cohort was derived from the multicenter AMLCG-1999 trial of the German AML Cooperative Group (GSE12417) with 163 primarily untreated CN-AML patients (median age = 57.5 years, range = 17–83 years). This cohort was used to validate the results of the first cohort.

All the clinical, cytogenetic, molecular, and microarray data of these CN-AML patients were obtained from the Gene Expression Omnibus (GEO) database1. Overall survival (OS) and event-free survival (EFS) data were obtained from the GEO database and their publications.

Microarray Analysis

Gene expression was derived from five GEO datasets—GSE9476, GSE30029, GSE6891, GSE12417, and GSE71014—using Affymetrix Human Genome 133 plus 2.0 and U133A gene chips. All datasets were normalized as previously described (16). Profiles of the messenger RNAs (mRNAs) and microRNAs and the genome-wide methylation data of 73 CN-AML patients were derived from The Cancer Genome Atlas (TCGA). Expression data of mRNA and microRNA were obtained by RNA sequencing, whereas methylation data was obtained by Illumina Infinium 450K BeadChips. One hundred and eighty-five CN-AML patients from GSE6891 (435 AML patients, no M3) were divided into four quartiles to choose the appropriate subdivision cutoff value. The expression of CLIC4 represented a normal distribution. A significant distinction was shown along the median value (Supplementary Figures S1A–C). Thus, the median value of CLIC4 expression was used to divide patients into CLIC4high and CLIC4low groups. The expression levels of ERG, DNMT3A, and other genes were examined using the same strategy.

Statistical Analysis

The primary endpoints were OS and EFS. The former was defined as the time from diagnosis to death by any cause, while the latter was defined as the time from date of diagnosis to the first event including failure to achieve complete remission (CR), relapse, or death. Between-group comparisons of OS and EFS were performed by the Kaplan–Meier and log-rank tests. The association of CLIC4 expression and clinical factors including age, sex, and FAB classification was analyzed using Fisher’s exact and Wilcoxon rank-sum tests for categorical and continuous variables, respectively. The impacts of CLIC4 expression on OS and EFS were assessed by multivariable hazard models. Multiple hypothesis correction (false discovery rate, FDR) as well as Student’s t-test were used to validate differences in the mRNA, microRNA, and DNA methylation profiles between the CLIC4high and CLIC4low groups (17). All analyses were performed by R3.6.3 and its related software packages.

Transcriptome, Gene Ontology, and Functional Enrichment Analysis

The differentially expressed genes analyzed in the genome-wide gene-expression profiles were then exported to ToppGene Suite2 (18). Functional enrichment analysis was performed using ToppGene Suite, which is widely accepted for gene ontology enrichment analysis. Hypergeometric distribution with Bonferroni correction was used as the standard method to determine significance. A FDR < 0.05 was considered to be significantly enriched for these analyses (19).

Weighted Correlation Network Analysis for Multi-Omics Data

Weighted correlation network analysis (WGCNA), one of the popular gene co-expression network identification tools (20), was adopted to further unveil the CLIC4-associated biomarkers across the mRNA, microRNA, and DNA methylation profiles. Gene filtering was performed with the TSUNAMI web portal (21). The target gene co-expression module, where CLIC4 was located at, was performed with downstream enrichment analysis.

Results

Overexpression of CLIC4 in AML Patients

Two public GEO microarray datasets (GSE9476 and GSE30029) were used to compare CILC4 expression in the BM, peripheral blood (PB), and CD34+ cell samples from AML patients and healthy donors. The results showed that CLIC4 was significantly overexpressed in AML BM than in normal BM (seven AML BM vs. 10 normal BM, GSE9476, P = 0.045) (Figure 1A), which was validated by PB in the same assay (19 AML PB vs. 10 normal PB, P = 0.0005) (Figure 1B). CD34+ cells derived from AML patients further validated CLIC4 overexpression in AML (47 AML CD34+ vs. 31 normal CD34+, GSE30029, P = 0.04) (Figure 1C). All results revealed that CLIC4 expression was markedly upregulated in AML patients.

FIGURE 1
www.frontiersin.org

Figure 1. Different expressions of CLIC4. (A) AML bone marrow samples (n = 7) vs. normal bone marrow samples (n = 10). (B) AML peripheral blood samples (n = 19) vs. normal peripheral blood samples (n = 10). (C) AML CD34+ samples (n = 47) and normal CD34+ samples (n = 31). AML, acute myeloid leukemia.

Clinical and Molecular Characteristics Between the CLIC4low and CLIC4high Groups in AML Patients

In the cohort of 185 CN-AML patients from the GSE6891 dataset, the CLIC4high group tends to have fewer NPM1 mutations (P = 0.0209) and more FLT3-ITD patients (P < 0.0001), while no significant differences between the CLIC4low and CLIC4high groups were observed in the CEBPA and N-ras/K-ras mutation groups (P = 0.5949 and P = 0.195, respectively). Furthermore, the CLIC4high group was more likely to overexpress BAALC, MAPKBP1, RUNX1, and TCF4, which were proven to contribute to adverse outcomes in CN-AML. Interestingly, patients in the CLIC4high group were more likely to relapse than those in the CLIC4low group (Table 1). These data indicated that CLIC4 overexpression was possibly related to adverse outcomes in CN-AML.

TABLE 1
www.frontiersin.org

Table 1. Clinical and molecular characteristics of 185 cytogenetically normal acute myeloid leukemia (CN-AML) patients in the CLIC4low and CLIC4high groups.

Overexpression of CLIC4 Was Associated With Unfavorable Outcomes in AML Patients

In order to further validate the prognostic role of CLIC4, survival analyses were carried out in the cohort of 185 CN-AML patients and 115 ELN Intermediate-I patients derived from GSE6891. The results showed that the CLIC4high group had notably shorter OS and EFS in CN-AML patients (CLIC4low vs. CLIC4high: median OS = 59.4 vs. 11.5 months, P < 0.0001; median EFS: 23.95 vs. 7.56 months, P < 0.0001) (Figures 2A,B) as well as in the ELN Intermediate-I category (CLIC4low vs. CLIC4high: median OS = 30.39 vs. 8.9 months, P = 0.015; median EFS = 13.01 vs. 6.83 months, P = 0.012) (Figures 2C,D). To further examine the impact of CLIC4 expression on survival, multivariable Cox proportional hazard models were constructed using multiple variables in the above cohorts (Table 2). For the cohort of 185 CN-AML patients, the CLIC4high group had 1.3 times higher risk of EFS (P < 0.01) and 1.36 higher risk of OS (P < 0.01). Other adverse factors included the presence of FLT3-ITD (P < 0.01 for both EFS and OS), the presence of FLT3-TKD (P < 0.01 for OS), and mutated N-ras/K-ras (P < 0.05 for EFS). For the cohort of 115 ELN Intermediate-I patients, the CLIC4high group had 1.23 times higher risk of EFS (P < 0.05) and 1.31 higher risk of OS (P < 0.01). Other adverse factors included the presence of FLT3-TKD (P < 0.01 for OS) and mutated IDH1/IDH2 (P < 0.05 for OS). These results indicated that the overexpression of CLIC4 was an independent risk factor for both OS and EFS in CN-AML patients and in the ELN Intermediate-I category.

FIGURE 2
www.frontiersin.org

Figure 2. The prognostic significance of CLIC4 expression in acute myeloid leukemia (AML) patients. Overall survival and event-free survival in 185 CN-AML patients (A,B), 115 ELN Intermediate-I patients (C,D), 255 NCCN-Intermediate Risk AML patients (E,F), and 435 non-M3 AML patients (G,H).

TABLE 2
www.frontiersin.org

Table 2. Multivariable analysis of overall survival (OS) and event-free survival (EFS) in 185 cytogenetically normal acute myeloid leukemia (CN-AML) and 115 European LeukemiaNet (ELN) Intermediate-I patients.

Additional survival analyses were carried out in the cohort of 255 NCCN Intermediate Risk patients and 435 non-M3 AML patients. Similarly, shorter OS and EFS were shown in the CLIC4high group both in the NCCN Intermediate Risk category (CLIC4low vs. CLIC4high: median OS = 50.4 vs. 13.06 months, P < 0.0001; median EFS = 21.85 vs. 8.35 months, P < 0.0001) (Figures 2E,F) and in non-M3 AML patients (CLIC4low vs. CLIC4high: median OS = 43.6 vs. 14.49 months, P < 0.0001; median EFS = 16.66 vs. 7.69 months, P < 0.0001) (Figures 2G,H). Another independent cohort of 163 de novo CN-AML patients (GSE12417) was further studied, which showed that CLIC4high was significantly associated with shorter OS (CLIC4low vs. CLIC4high: median OS = 14.73 vs. 11 months, P = 0.0032) (Supplementary Figure S2).

Genome-Wide Gene Expression Profiles Associated With CLIC4 Expression

To gain a better understanding of the biological value of CLIC4 in leukemogenesis, we derived the CLIC4-associated gene expression profiles from high-throughput sequencing in the 73 CN-AML patients from the TCGA dataset. A total of 1,185 upregulated 538 downregulated genes were found to be significantly associated with the CLIC4high group (FDR-adjusted P < 0.01) (Figures 3A,B). The 1,185 upregulated genes were further exported to the ToppGene Suite. A total of 105 genes were found to be significantly enriched in chronic lymphocytic leukemia (CLL) (overlap = 105/633, P = 6.461e-32) (Supplementary Table S1) (22). Another set of 153 genes were found to be enriched in CD34+ cells isolated from the BM of patients with chronic myeloid leukemia (CML) compared to those from normal donors (overlap = 153/1,399, P = 2.400e-25) (Supplementary Table S2) (23). Besides, a total of 53 genes were significantly upregulated in cancer stem cells isolated from mammary tumors compared to those from the non-tumorigenic cells (overlap = 53/432, P = 3.249e-11) (Supplementary Table S3) (24). Interestingly, 19 genes were significantly enriched in a probe-set gene-expression signature that predicted survival in CN-AML (overlap = 19/62, P = 1.406e-11) (Supplementary Table S4) (16). The top Gene Ontology (GO) biological process term was protein ubiquitination (GO:0016567; overlap = 69/852, P = 4.282e-5). Five hundred and thirty-eight downregulated genes were analyzed as previously described. A total of 23 genes were among the top 100 probe sets for pediatric AML subtypes with chimeric MLL fusions (overlap = 23/81, P = 1.033e-19) (Supplementary Table S5). The top GO biological process terms were myeloid cell activation involved in immune response (GO:0002275; overlap = 82/557, P = 1.563e-40) and myeloid leukocyte-mediated immunity (GO:0002444; overlap = 82/568, P = 7.022e-40).

FIGURE 3
www.frontiersin.org

Figure 3. Genome-wide genes/cell signaling pathways associated with CLIC4 expression. (A) Volcano plot of the genome-wide gene profiles between the CLIC4high and CLIC4low groups. (B) Gene expression heatmap. The top curve indicates the expression distributions of CLIC4 in 73 cytogenetically normal acute myeloid leukemia (CN-AML) samples. (C) Cell signaling pathways associated with CLIC4 expression.

Cell signal pathways associated with CLIC4, analyzed from the MSigDB data, further provided new insights into the leukemogenesis of CN-AML. A total of five upregulated and 17 downregulated pathways showed significant associations with the CLIC4high group (P < 0.05; Figure 3C). The upregulated pathway included the hippo signaling pathway, an important canonical oncogenic pathway. However, the downregulated pathways were mostly immune activation pathways such as natural killer cell-mediated cytotoxicity, as well as chemokine and neurotrophin signaling pathways. These dysregulated gene expressions and signaling pathways might provide insights into the involvement between CLIC4 and leukemogenesis of CN-AML.

Genome-Wide MicroRNA Profiles Associated With CLIC4 Expression

Investigation of the dysregulated microRNAs could be used to determine the extent of heterogeneity in AML. Therefore, the CLIC4-associated genome-wide microRNA profiles in 73 CN-AML patients from the TCGA dataset were analyzed. A total of 67 microRNAs were significantly correlated with CLIC4 expression, including 62 positive and five negative microRNAs (P < 0.05; Figures 4A,B). Positively correlated microRNAs included miR-25, miR-106b, miR-146a, miR-146b, miR-155, miR-181a-1, miR-181a-2, miR-181b-1, miR-181b-2, miR-181c, miR-181d, etc. MiR-25 was found to be overexpressed in the c-kit subgroup and involved in leukemogenesis in pediatric AML. MiR-106b was significantly upregulated in refractory and relapsed pediatric MLL-rearranged AML (25). MiR-146a and miR-146b were abundant in t(8;21) AML, while miR-155 were abundant in FLT3-ITD AML (26). The miR-181 family (miR-181a-1, miR-181a-2, miR-181b-1, miR-181b-2, miR-181c, and miR-181d) is regarded as oncogenic in several cancers and its upregulated expression predicted unfavorable outcomes in AML (27). The negatively correlated microRNAs were miR-10a, miR-146b, miR-1266, etc. MiR-10a was found downregulated in hematological tumor cell lines by targeting BCL6 (28). MiR-146b could delay the aggressiveness and progression of T-ALL (29). miR-1266 was downregulated in childhood T-ALL and might be used to distinguish childhood ALL subtypes (30).

FIGURE 4
www.frontiersin.org

Figure 4. Interaction network analysis of genome-wide microRNAs and their target genes associated with CLIC4 expression. (A) Volcano plot of the genome-wide microRNA profiles between the CLIC4high and CLIC4low groups. (B) MicroRNA expression heatmap. The top curve indicates the expression distributions of CLIC4 in 73 cytogenetically normal acute myeloid leukemia (CN-AML) samples. (C) MicroRNA–mRNA interaction network analysis (red: upregulation; green: downregulation; rectangle: microRNAs; ellipse: mRNAs).

Interaction network analysis was conducted to further validate the microRNA–target gene correlation results (Figure 4C). Many tumor repressors were found to be the targets of upregulated microRNAs, contributing to worse outcomes. For example, GPR132, targeted by miR-146b, could exert growth inhibitory and apoptotic effects in AML, ALL, and CML cell lines (31). PYCARD, targeted by miR-125a, was shown to be downregulated to different extents in a wide spectrum of human cancers and prevented tumor cells from undergoing apoptosis (32). These results helped explain the reason why CLIC4 acted as an adverse biomarker and provided a comprehensive view of its molecular mechanisms.

Genome-Wide Methylation Profiles Associated With CLIC4 Expression

Differential methylated regions (DMR) were further derived to investigate the different methylation patterns between the CLIC4low and CLIC4high groups in 73 CN-AML patients from the TCGA dataset. Firstly, a total of 265 hypomethylation and 675 hypermethylation DMRs were derived from the comparison between the CLIC4low and CLIC4high groups [P < 0.05, | log2(FC)| > 1] (Figure 5A). Secondly, position distribution around the CpG islands was compared in these abnormal DMRs. More hypermethylation DMRs were around the CpG islands (Island: P = 1.22e-12, N_Shore: P = 2.61e-02) and in open sea regions (S_Shelf: P = 1.29e-02, N_Shelf: P = 8.81e-03) (upper subfigure of Figure 5B). Thirdly, the position distribution of the different gene structural fragments was presented, showing hypermethylated DMRs lying on TSS1500, gene body, the 5′UTR region, 3′UTR region, and first exon (P = 1.39e-03, P = 3.75e-03, P = 4.66e-04, P = 2.99e-05, and P = 7.04e-03, respectively) (lower subfigure of Figure 5B). Finally, CLIC4-related DMRs enriched on islands and gene promoter regions were presented in heatmaps (Figures 5C,D).

FIGURE 5
www.frontiersin.org

Figure 5. Genome-wide methylation profiles associated with CLIC4 expression. (A) A total of 265 hypomethylation and 675 hypermethylation differential methylated regions were discovered from the comparison between the CLIC4high and CLIC4low groups. (B) More hypermethylation differential methylated regions were found around the CpG islands and in open sea regions, while more hypermethylated differential methylated regions lie on the body and the 3′UTR region. (C,D) Heatmaps showed CLIC4-associated differential methylated regions enriched on islands and gene promoter regions.

Identifying CLIC4-Associated Biomarkers With Multi-Omics WGCNA

As the genomics and transcriptomics data remain noises, it is necessary to filter out some genes before the multi-omics co-expression analysis by WGCNA. We filtered out mRNA genes with the lowest 20% means and variances of expression values, filtered out microRNA genes with the lowest 20% means and variances of expression values, and filtered out methylation profiles with the lowest 80% means and variances of expression values via the TSUNAMI web portal. We ended up with 13,082 mRNA genes, 681 microRNA genes, and 15,843 methylation profiles. We considered them as the combined multi-omics biomarkers (29,606 in total) and performed biomarker-wise normalization (norm = 1 for all biomarkers) before performing the WGCNA.

By selecting the soft threshold (power) = 6 (Figure 6A), minimum module size = 10, and merge cut height of the dendrogram = 0.25, 136 co-expressed modules were identified (Figure 6B). The gene of interest CLIC4 is located at the module with a dark turquoise color which contained 25 mRNA genes, five microRNAs (miR-211, miR-28, miR-511-1, miR-511-2, and miR-581), and 238 methylation profiles. The complete co-expression module list is provided in a Supplementary Material.

FIGURE 6
www.frontiersin.org

Figure 6. Results of the multi-omics analysis with weighted correlation network analysis (WGCNA). (A) Scale-free topology plot to determine soft threshold (power). (B) The identified 136 WGCNA co-expression modules and the dendrogram.

Enrichment analysis was further performed based on the targeted co-expression module with 25 mRNA genes (include CLIC4). We found that those genes were enriched in the chromosome location 19p13.3 (genes: SEMA6B, LRG1, and PLIN5; overlap = 3/25, P = 5.698e-4), in agreement with several existing literatures (33, 34), which further confirmed the module relationship to the AML.

Discussion

With the increasing use of multi-omics analysis in research and clinical routine, the current prognostic genes in CN-AML will likely be expanded. The new gene list will serve as a valuable tool in advancing the individualized treatment approaches in CN-AML. Recent studies has shown that CLIC4, an integral component of TGF-β signaling, has been proven to either enhance or inhibit tumor growth depending on the tumor type. Downregulation of CLIC4 could promote tumor cell growth and clonogenicity in lung cancer and gastric cancer, while its upregulation could cause tumor cell growth in squamous cell carcinomas and breast cancer (35).

Our study demonstrated a critical unfavorable prognostic role of CLIC4 for CN-AML. Firstly, CLIC4 was found overexpressed in AML BM and PB samples than in normal BM and PB samples, and also in AML CD34+ than in normal CD34+ samples (Figure 1). Secondly, the clinical and molecular characteristics related to CLIC4 expression were explored in a cohort of 185 CN-AML patients. We found that the CLIC4high group contained fewer NPM1 mutations (P = 0.0209) and more FLT3-ITD patients (P < 0.0001) and remarkably high expressions of BAALC (P = 0.0153), MAPKBP1 (P = 0.0153), RUNX1 (P = 0.0026), and TCF4 (P < 0.0001), which were proven to contribute to unfavorable outcomes in CN-AML. The CLIC4high group also had more relapse patients than the CLIC4low group (Table 1). Multivariable Cox proportional hazard models further indicated CLIC4 as an independent biomarker for CN-AML (Table 2). Thirdly, survival analysis based on two relatively large and independent CN-AML cohorts (GSE6891 and GSE12417) showed that CLIC4high was associated with adverse outcomes (Figures 2A–D). Similar results were shown in ELN Intermediate-I category, NCCN Intermediate Risk category, and non-M3 AML patients (GSE6891; Figures 2E–H). Our results might provide insights into further stratification of CN-AML, ELN Intermediate-I category, and NCCN Intermediate Risk category.

Multi-omics analysis of the genetic and epigenetic mechanisms was important in leukemogenesis, including abnormal gene expression, microRNA–target gene interaction network analysis, and downstream gene ontology analysis (18, 36). Our study further analyzed how CLIC4 overexpression affected the prognosis in CN-AML from these aspects. We derived the CLIC4-associated genes, microRNAs, and cell signaling pathways. Using the ToppGene Suite, we discovered several sets of genes that were significantly co-expressed in CLL, CML, cancer stem cells, as well as probe-set signature that predicted survival in CN-AML. Dysregulated signaling pathways also provided evidence of the involvement of CLIC4 in multiple cellular pathways (Figure 3).

MicroRNA-involved mechanisms related to the prognostic role of CLIC4 were further studied. Those that were known as oncogenic or tumor suppressive were positively or negatively correlated with CLIC4, respectively (such as miR-181 family and miR-10a). Other synchronized changes of microRNA–target gene (such as GPR132 targeted by miR-146b and PYCARD targeted by miR-125a) might promote leukemogenesis in CN-AML (Figure 4). DMRs were mostly hypermethylated around the CpG islands, in open sea regions, and in almost all gene structural fragments. Heatmaps of the CLIC4-related DMRs enriched on islands and gene promoter regions were also presented (Figure 5). These genes, pathways, microRNAs, microRNA–target genes, and hypermethylation DMRs might be associated with adverse prognosis of CN-AML and establish a better understanding of leukemogenesis.

Nevertheless, a multi-omics WGCNA was further performed by combining the mRNAs, microRNAs, and methylation profiles. The targeted module which contains CLIC4 was also genetically enriched in chromosome location 19p13.3, which not only reflects some existing leukemia literatures but also suggests a potential important cytoband which deserves further analysis.

Conclusion

In summary, we performed a large-scale multi-omics analysis of CLIC4 in CN-AML and portrayed a hypothetical mechanism figure (Supplementary Figure S3). We provided strong evidences that CLIC4high patients were characterized by different adverse factors and that overexpression of CLIC4 was an adverse biomarker for CN-AML. Novel targeted therapeutic approaches and more prognostic biomarkers will be further discovered, which hold promise for improving CN-AML patients’ outcomes.

Data Availability Statement

Publicly available datasets were analyzed in this study. This data can be found here: Gene Expression Omnibus (GSE9476, GSE30029, GSE6891, GSE12417, and GSE71014) and The Cancer Genome Atlas.

Author Contributions

SH, ZH, and PC designed and performed the study and wrote the manuscript. ZH and CF analyzed the data and revised the manuscript. All authors reviewed the final manuscript.

Funding

The present study was supported by grants from the National Natural Science Foundation of China (81701961), the Beijing Natural Science Foundation (7204305), the Beijing Science and Technology New Star Project (XX2018019/Z181100006218028), and the PLA General Hospital Science and Technology Project (18KMM01).

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Supplementary Material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fonc.2020.01648/full#supplementary-material

FIGURE S1 | Median value of CLIC4 expression as the cut-off. (A) Normally distribution of CLIC4 expression. (B) OS and (C) EFS of CN-AML patients, the patients were subdivided into four quartiles based on the quartile of CLIC4 expression.

FIGURE S2 | The validation of the unfavorable outcome associated with CLIC4high in a cohort of 163 CN-AML patients.

FIGURE S3 | The hypothetical mechanism figure of CLIC4 in CN-AML.

Footnotes

  1. ^ http://www.ncbi.nlm.nih.gov/geo
  2. ^ https://toppgene.cchmc.org/

References

1. Arindrarto W, Borràs DM, de Groen RAL, van den Berg RR, Locher IJ, van Diessen S, et al. Comprehensive diagnostics of acute myeloid leukemia by whole transcriptome RNA sequencing. Leukemia. (2020). doi: 10.1038/s41375-020-0762-8 [Epub ahead of print].

CrossRef Full Text | PubMed Abstract | Google Scholar

2. Lin SY, Hu FF, Miao YR, Hu H, Lei Q, Zhang Q, et al. Identification of STAB1 in multiple datasets as a prognostic factor for cytogenetically normal AML: mechanism and drug indications. Mol Ther Nucleic Acids. (2019) 18:476–84. doi: 10.1016/j.omtn.2019.09.014

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Wang M, Yang C, Zhang L, Schaar DG. Molecular mutations and their cooccurrences in cytogenetically normal acute myeloid leukemia. Stem Cells Int. (2017) 2017:6962379. doi: 10.1155/2017/6962379

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Zhou YL, Wu LX, Peter Gale R, Wang ZL, Li JL, Jiang H, et al. Mutation topography and risk stratification for de novo acute myeloid leukaemia with normal cytogenetics and no nucleophosmin 1 (NPM1) mutation or Fms-like tyrosine kinase 3 internal tandem duplication (FLT3-ITD). Br J Haematol. (2020) 190:274–83. doi: 10.1111/bjh.16526

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Döhner H, Estey E, Grimwade D, Amadori S, Appelbaum FR, Büchner T, et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. (2017) 129:424–47. doi: 10.1182/blood-2016-08-733196

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Arcangeli A, Becchetti A. Novel perspectives in cancer therapy: targeting ion channels. Drug Resist Updat. (2015) 21–22:11–9. doi: 10.1016/j.drup.2015.06.002

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Rafieemehr H, Samimi A, Maleki Behzad M, Ghanavat M, Shahrabi S. Altered expression and functional role of ion channels in leukemia: bench to bedside. Clin Transl Oncol. (2020) 22:283–93. doi: 10.1007/s12094-019-02147-2

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Prevarskaya N, Skryma R, Shuba Y. Ion channels and the hallmarks of cancer. Trends Mol Med. (2010) 16:107–21. doi: 10.1016/j.molmed.2010.01.005

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Chou SY, Hsu KS, Otsu W, Hsu YC, Luo YC, Yeh C, et al. CLIC4 regulates apical exocytosis and renal tube luminogenesis through retromer- and actin-mediated endocytic trafficking. Nat Commun. (2016) 7:10412. doi: 10.1038/ncomms10412

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Malik M, Jividen K, Padmakumar VC, Cataisson C, Li L, Lee J, et al. Inducible NOS-induced chloride intracellular channel 4 (CLIC4) nuclear translocation regulates macrophage deactivation. Proc Natl Acad Sci USA. (2012) 109:6130–5. doi: 10.1073/pnas.1201351109

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Abdul-Salam VB, Russomanno G, Chien-Nien C, Mahomed AS, Yates LA, Wilkins MR, et al. CLIC4/Arf6 pathway. Circ Res. (2019) 124:52–65. doi: 10.1161/circresaha.118.313705

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Wojciak-Stothard B, Abdul-Salam VB, Lao KH, Tsang H, Irwin DC, Lisk C, et al. Aberrant chloride intracellular channel 4 expression contributes to endothelial dysfunction in pulmonary arterial hypertension. Circulation. (2014) 129:1770–80. doi: 10.1161/circulationaha.113.006797

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Deng YJ, Tang N, Liu C, Zhang JY, An SL, Peng YL, et al. CLIC4, ERp29, and Smac/DIABLO derived from metastatic cancer stem-like cells stratify prognostic risks of colorectal cancer. Clin Cancer Res. (2014) 20:3809–17. doi: 10.1158/1078-0432.Ccr-13-1887

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Liu G, Zhao Y, Chen H, Jia J, Cheng X, Wang F, et al. Analysis of differentially expressed genes in a Chinese cohort of esophageal squamous cell carcinoma. J Cancer. (2020) 11:3783–93. doi: 10.7150/jca.40850

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Peretti M, Angelini M, Savalli N, Florio T, Yuspa SH, Mazzanti M. Chloride channels in cancer: focus on chloride intracellular channel 1 and 4 (CLIC1 AND CLIC4) proteins in tumor development and as novel therapeutic targets. Biochim Biophys Acta. (2015) 1848:2523–31. doi: 10.1016/j.bbamem.2014.12.012

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Metzeler KH, Hummel M, Bloomfield CD, Spiekermann K, Braess J, Sauerland MC, et al. An 86-probe-set gene-expression signature predicts survival in cytogenetically normal acute myeloid leukemia. Blood. (2008) 112:4193–201. doi: 10.1182/blood-2008-02-134411

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Hanniford D, Segura MF, Zhong J, Philips E, Jirau-Serrano X, Darvishian F, et al. Identification of metastasis-suppressive microRNAs in primary melanoma. J Natl Cancer Inst. (2015) 107:dju494. doi: 10.1093/jnci/dju494

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Huang Z, Zhan X, Xiang S, Johnson TS, Helm B, Yu CY, et al. SALMON: survival analysis learning with multi-omics neural networks on breast cancer. Front Genet. (2019) 10:166. doi: 10.3389/fgene.2019.00166

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Chen J, Bardes EE, Aronow BJ, Jegga AG. ToppGene suite for gene list enrichment analysis and candidate gene prioritization. Nucleic Acids Res. (2009) 37:W305–11. doi: 10.1093/nar/gkp427

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Langfelder P, Horvath S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinform. (2008) 9:559. doi: 10.1186/1471-2105-9-559

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Huang Z, Han Z, Wang TX, Shao W. TSUNAMI: translational bioinformatics tool suite for network analysis and mining. bioRxiv. (2019). [Preprint]. doi: 10.1101/787507

CrossRef Full Text | Google Scholar

22. Calin GA, Cimmino A, Fabbri M, Ferracin M, Wojcik SE, Shimizu M, et al. MiR-15a and miR-16-1 cluster functions in human leukemia. Proc Natl Acad Sci USA. (2008) 105:5166–71. doi: 10.1073/pnas.0800121105

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Diaz-Blanco E, Bruns I, Neumann F, Fischer JC, Graef T, Rosskopf M, et al. Molecular signature of CD34(+) hematopoietic stem and progenitor cells of patients with CML in chronic phase. Leukemia. (2007) 21:494–504. doi: 10.1038/sj.leu.2404549

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Zhang M, Behbod F, Atkinson RL, Landis MD, Kittrell F, Edwards D, et al. Identification of tumor-initiating cells in a p53-null mouse model of breast cancer. Cancer Res. (2008) 68:4674–82. doi: 10.1158/0008-5472.Can-07-6353

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Huang S, Huang Z, Ma C, Luo L, Li YF, Wu YL, et al. Acidic leucine-rich nuclear phosphoprotein-32A expression contributes to adverse outcome in acute myeloid leukemia. Ann Transl Med. (2020) 8:345. doi: 10.21037/atm.2020.02.54

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Lim EL, Trinh DL, Ries RE, Wang J, Gerbing RB, Ma Y, et al. MicroRNA expression-based model indicates event-free survival in pediatric acute myeloid leukemia. J Clin Oncol. (2017) 35:3964–77. doi: 10.1200/jco.2017.74.7451

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Pop-Bica C, Pintea S, Cojocneanu-Petric R, Del Sal G, Piazza S, Wu ZH, et al. MiR-181 family-specific behavior in different cancers: a meta-analysis view. Cancer Metastasis Rev. (2018) 37:17–32. doi: 10.1007/s10555-017-9714-9

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Fan Q, Meng X, Liang H, Zhang H, Liu X, Li L, et al. miR-10a inhibits cell proliferation and promotes cell apoptosis by targeting BCL6 in diffuse large B-cell lymphoma. Protein Cell. (2016) 7:899–912. doi: 10.1007/s13238-016-0316-z

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Correia NC, Fragoso R, Carvalho T, Enguita FJ, Barata JT. MiR-146b negatively regulates migration and delays progression of T-cell acute lymphoblastic leukemia. Sci Rep. (2016) 6:31894. doi: 10.1038/srep31894

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Almeida RS, Costa ESM, Coutinho LL, Garcia Gomes R, Pedrosa F, Massaro JD, et al. MicroRNA expression profiles discriminate childhood T- from B-acute lymphoblastic leukemia. Hematol Oncol. (2019) 37:103–12. doi: 10.1002/hon.2567

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Nii T, Prabhu VV, Ruvolo V, Madhukar N, Zhao R, Mu H, et al. Imipridone ONC212 activates orphan G protein-coupled receptor GPR132 and integrated stress response in acute myeloid leukemia. Leukemia. (2019) 33:2805–16. doi: 10.1038/s41375-019-0491-z

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Protti MP, De Monte L. Dual role of inflammasome adaptor ASC in cancer. Front Cell Dev Biol. (2020) 8:40. doi: 10.3389/fcell.2020.00040

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Taniwaki M, Nishida K, Takashima T, Nakagawa H, Fujii H, Tamaki T, et al. Nonrandom chromosomal rearrangements of 14q32.3 and 19p13.3 and preferential deletion of 1p in 21 patients with multiple myeloma and plasma cell leukemia. Blood. (1994) 84:2283–90.

Google Scholar

34. Barber KE, Harrison CJ, Broadfield ZJ, Stewart AR, Wright SL, Martineau M, et al. Molecular cytogenetic characterization of TCF3 (E2A)/19p13.3 rearrangements in B-cell precursor acute lymphoblastic leukemia. Genes Chromosomes Cancer. (2007) 46:478–86. doi: 10.1002/gcc.20431

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Wang B, Zheng J, Chen Q, Wu C, Li Y, Yu XY, et al. CLIC4 abrogation promotes epithelial mesenchymal transition in gastric cancer. Carcinogenesis. (2019) 41:841–9. doi: 10.1093/carcin/bgz156

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Huang Z, Johnson TS, Han Z, Helm B, Cao S, Zhang C, et al. Deep learning-based cancer survival prognosis from RNA-seq data: approaches and evaluations. BMC Med Genomics. (2020) 13:41. doi: 10.1186/s12920-020-0686-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: CLIC4, expression, prognostic biomarker, CN-AML, adverse outcome

Citation: Huang S, Huang Z, Chen P and Feng C (2020) Aberrant Chloride Intracellular Channel 4 Expression Is Associated With Adverse Outcome in Cytogenetically Normal Acute Myeloid Leukemia. Front. Oncol. 10:1648. doi: 10.3389/fonc.2020.01648

Received: 01 July 2020; Accepted: 27 July 2020;
Published: 09 September 2020.

Edited by:

Basem M. William, The Ohio State University, United States

Reviewed by:

Michael Poidinger, Royal Children’s Hospital, Australia
Tanima Bose, Ludwig Maximilian University of Munich, Germany

Copyright © 2020 Huang, Huang, Chen and Feng. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Cong Feng, congf@mit.edu; Sai Huang, helinahs@foxmail.com

These authors have contributed equally to this work

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.